scispace - formally typeset
Search or ask a question
Journal ArticleDOI

70-Gene Signature as an Aid to Treatment Decisions in Early-Stage Breast Cancer

TL;DR: Among women with early-stage breast cancer who were at high clinical risk and low genomic risk for recurrence, the receipt of no chemotherapy on the basis of the 70-gene signature led to a 5-year rate of survival without distant metastasis that was 1.5 percentage points lower than the rate with chemotherapy.
Abstract: BackgroundThe 70-gene signature test (MammaPrint) has been shown to improve prediction of clinical outcome in women with early-stage breast cancer. We sought to provide prospective evidence of the clinical utility of the addition of the 70-gene signature to standard clinical–pathological criteria in selecting patients for adjuvant chemotherapy. MethodsIn this randomized, phase 3 study, we enrolled 6693 women with early-stage breast cancer and determined their genomic risk (using the 70-gene signature) and their clinical risk (using a modified version of Adjuvant! Online). Women at low clinical and genomic risk did not receive chemotherapy, whereas those at high clinical and genomic risk did receive such therapy. In patients with discordant risk results, either the genomic risk or the clinical risk was used to determine the use of chemotherapy. The primary goal was to assess whether, among patients with high-risk clinical features and a low-risk gene-expression profile who did not receive chemotherapy, the...

Summary (1 min read)

Jump to: [BACKGROUND][METHODS][RESULTS] and [CONCLUSIONS]

BACKGROUND

  • The 70-gene signature test has been shown to improve prediction of clinical outcome in women with early-stage breast cancer.
  • The authors sought to provide prospective evidence of the clinical utility of the addition of the 70-gene signature to standard clinical–pathological criteria in selecting patients for adjuvant chemotherapy.

METHODS

  • Phase 3 study, the authors enrolled 6693 women with early-stage breast cancer and determined their genomic risk (using the 70-gene signature) and their clinical risk (using a modified version of Adjuvant!.
  • Women at low clinical and genomic risk did not receive chemotherapy, whereas those at high clinical and genomic risk did receive such therapy.
  • In patients with discordant risk results, either the genomic risk or the clinical risk was used to determine the use of chemotherapy.
  • The primary goal was to assess whether, among patients with high-risk clinical features and a low-risk gene-expression profile who did not receive chemotherapy, the lower boundary of the 95% confidence interval for the rate of 5-year survival without distant metastasis would be 92% (i.e., the noninferiority boundary) or higher.

RESULTS

  • At 5 years, the rate of survival without distant metastasis in this group was 94.7% (95% confidence interval, 92.5 to 96.2) among those not receiving chemotherapy.
  • The absolute difference in this survival rate between these patients and those who received chemother apy was 1.5 percentage points, with the rate being lower without chemotherapy.
  • Similar rates of survival without distant metastasis were reported in the subgroup of patients who had estrogen-receptor–positive, human epidermal growth factor receptor 2–negative, and either node-negative or node-positive disease.

CONCLUSIONS

  • Among women with early-stage breast cancer who were at high clinical risk and low genomic risk for recurrence, the receipt of no chemotherapy on the basis of the 70-gene signature led to a 5-year rate of survival without distant metastasis that was 1.5 percentage points lower than the rate with chemotherapy.
  • The New England Journal of Medicine Downloaded from nejm.org at IMPERIAL COLLEGE LONDON on September 1, 2016.
  • Treatment decisions are based on characteristics of the tumor (hormonal receptor and HER2 status, tumor grade and size, and lymph-node status) and of the patient (age, menopausal status, and performance status).
  • Online2,3 and PREDICT Plus,4 were created to assist in such decision making.

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

The new england
journal of medicine
n engl j med 375;8 nejm.org August 25, 2016
717
established in 1812
August 25, 2016
vol. 375 no. 8
The authors’ full names, academic de-
grees, and affiliations are listed in the
Appendix. Address reprint requests to
Dr. Cardoso at the Breast Unit, Champali-
maud Clinical Center–Champalimaud
Foundation, Lisbon, Portugal, or at
mindact@ eortc . be.
* A complete list of the Microarray in
Node-Negative and 1 to 3 Positive Lymph
Node Disease May Avoid Chemotherapy
(MINDACT) investigators is provided in
the Supplementary Appendix, available
at NEJM.org.
Drs. Cardoso, van’t Veer, and Bogaerts
and Drs. Rutgers and Piccart contributed
equally to this article.
This article was updated on August 25,
2016, at NEJM.org.
N Engl J Med 2016;375:717-29.
DOI: 10.1056/NEJMoa1602253
Copyright © 2016 Massachusetts Medical Society.
BACKGROUND
The 70-gene signature test (MammaPrint) has been shown to improve prediction of
clinical outcome in women with early-stage breast cancer. We sought to provide prospec-
tive evidence of the clinical utility of the addition of the 70-gene signature to standard
clinical–pathological criteria in selecting patients for adjuvant chemotherapy.
METHODS
In this randomized, phase 3 study, we enrolled 6693 women with early-stage breast cancer
and determined their genomic risk (using the 70-gene signature) and their clinical risk (using
a modified version of Adjuvant! Online). Women at low clinical and genomic risk did not re-
ceive chemotherapy, whereas those at high clinical and genomic risk did receive such therapy.
In patients with discordant risk results, either the genomic risk or the clinical risk was used
to determine the use of chemotherapy. The primary goal was to assess whether, among pa-
tients with high-risk clinical features and a low-risk gene-expression profile who did not re-
ceive chemotherapy, the lower boundary of the 95% confidence interval for the rate of 5-year
survival without distant metastasis would be 92% (i.e., the noninferiority boundary) or higher.
RESULTS
A total of 1550 patients (23.2%) were deemed to be at high clinical risk and low ge-
nomic risk. At 5 years, the rate of survival without distant metastasis in this group was
94.7% (95% confidence interval, 92.5 to 96.2) among those not receiving chemotherapy.
The absolute difference in this survival rate between these patients and those who re-
ceived chemother apy was 1.5 percentage points, with the rate being lower without chemo-
therapy. Similar rates of survival without distant metastasis were reported in the subgroup
of patients who had estrogen-receptor–positive, human epidermal growth factor receptor
2–negative, and either node-negative or node-positive disease.
CONCLUSIONS
Among women with early-stage breast cancer who were at high clinical risk and low
genomic risk for recurrence, the receipt of no chemotherapy on the basis of the 70-gene
signature led to a 5-year rate of survival without distant metastasis that was 1.5 percent-
age points lower than the rate with chemotherapy. Given these findings, approximately
46% of women with breast cancer who are at high clinical risk might not require chemo-
therapy. (Funded by the European Commission Sixth Framework Program and others;
ClinicalTrials.gov number, NCT00433589; EudraCT number, 2005-002625-31.)
abstract
70-Gene Signature as an Aid to Treatment Decisions
in Early-Stage Breast Cancer
F. Cardoso, L.J. van’t Veer, J. Bogaerts, L. Slaets, G. Viale, S. Delaloge, J.-Y. Pierga, E. Brain, S. Causeret,
M. DeLorenzi, A.M. Glas, V. Golfinopoulos, T. Goulioti, S. Knox, E. Matos, B. Meulemans, P.A. Neijenhuis, U. Nitz,
R. Passalacqua, P. Ravdin, I.T. Rubio, M. Saghatchian, T.J. Smilde, C. Sotiriou, L. Stork, C. Straehle, G. Thomas,
A.M. Thompson, J.M. van der Hoeven, P. Vuylsteke, R. Bernards, K. Tryfonidis, E. Rutgers, and M. Piccart,
for the MINDACT Investigators*
The New England Journal of Medicine
Downloaded from nejm.org at IMPERIAL COLLEGE LONDON on September 1, 2016. For personal use only. No other uses without permission.
Copyright © 2016 Massachusetts Medical Society. All rights reserved.

n engl j med 375;8 nejm.org August 25, 2016
718
The
new england journal
of
medicine
W
omen with early-stage breast
cancer are often treated with adjuvant
systemic therapy consisting of chemo-
therapy, endocrine therapy, agents against human
epidermal growth factor receptor 2 (HER2), or
combinations of these drugs when appropriate.
Treatment decisions are based on characteristics
of the tumor (hormonal receptor and HER2 status,
tumor grade and size, and lymph-node status) and
of the patient (age, menopausal status, and per-
formance status).
1
Tools that incorporate these
features, such as Adjuvant! Online
2,3
and PREDICT
Plus,
4
were created to assist in such decision
making. However, these algorithms do not take
into account the individual biologic characteris-
tics of the patients tumor.
5
A substantial num-
ber of patients with breast cancer are overtreated
and are thus being exposed to the risk of toxic
effects from adjuvant therapy without deriving
significant benefit. In 2007, respondents to an
international survey identified the use of mo-
lecular signatures to select patients who could
be spared adjuvant therapy as a high priority.
6
Gene-expression profiling studies have distin-
guished at least four molecularly distinct types of
breast cancer.
7,8
Several genomic tests have been
developed to better predict clinical outcome and
to determine whether the addition of adjuvant
chemotherapy to endocrine therapy is worth-
while.
8,9
One such test, the 70-gene signature
10
(MammaPrint), classifies tumors into groups
that are associated with a good prognosis or a
poor prognosis on the basis of the risk of distant
recurrence at 5 years and at 10 years.
11
An inde-
pendent validation study by the TRANSBIG con-
sortium, a network of some 40 partners in 21
countries associated with the Breast Interna-
tional Group (BIG), confirmed that the 70-gene
signature, which has been approved by the Food
and Drug Administration (FDA), is able to distin-
guish patients who are at significant risk for
distant relapse and death from those at low risk.
12
In this international, prospective, randomized,
phase 3 study, called the Microarray in Node-
Negative and 1 to 3 Positive Lymph Node Dis-
ease May Avoid Chemotherapy (EORTC 10041/
BIG 3-04 MINDACT) study, we sought to provide
evidence of the clinical utility
13-15
of the addition
of the 70-gene signature to standard clinical–
pathological criteria in selecting patients for adju-
vant chemotherapy.
16,17
Here, we report 5-year
outcomes and the results of the treatment ran-
domization for groups with discordance in risk.
Methods
Study Patients
From 2007 through 2011, we enrolled patients at
112 institutions in nine European countries. Eli-
gible patients were women between the ages of
18 and 70 years with histologically confirmed
primary invasive breast cancer (stage T1 or T2 or
operable T3). In the initial study design, all the
patients had to have lymph-node–negative dis-
ease, as described in the protocol, available with
the full text of this article at NEJM.org. As of
August 2009, the protocol was revised to allow
the enrollment of women with up to three posi-
tive axillary nodes.
18
The study design called for
following all the patients according to local
standards for at least 10 years; those receiving
endocrine therapy will be followed for a mini-
mum of 15 years.
Written informed consent was obtained from
all the patients. The protocol review committee
of the European Organization for Research and
Treatment of Cancer (EORTC) and the ethics
committee at each participating site approved
the study.
Prognostic Tools
We used the 70-gene signature to determine
genomic risk and Adjuvant! Online (version 8.0
with HER2 status) (www.adjuvantonline.com) to
determine clinical risk (Fig. S7 in the Supple-
mentary Appendix, available at NEJM.org). De-
tails regarding clinical risk assessment accord-
ing to the modified version of Adjuvant! Online
are provided in Table S13 in the Supplementary
Appendix.
A frozen sample of the resected tumor was
shipped on dry ice to Agendia for molecular
diagnostic testing. The quantification of the
tumor-cell percentage was followed by 70-gene
profiling embedded in a whole-transcriptome
array that is cleared for use by the FDA.
19,20
Categorization into Risk Groups
A low clinical risk was defined as the 10-year
probability of breast-cancerspecific survival with-
out systemic therapy of more than 88% among
women with estrogen receptor (ER)–positive
tumors and more than 92% among women with
ER-negative tumors, to account for the 4-percent-
age-point average absolute benefit of adjuvant
endocrine therapy for ER-positive tumors. Pa-
tients with low-risk disease according to both
The New England Journal of Medicine
Downloaded from nejm.org at IMPERIAL COLLEGE LONDON on September 1, 2016. For personal use only. No other uses without permission.
Copyright © 2016 Massachusetts Medical Society. All rights reserved.

n engl j med 375;8 nejm.org August 25, 2016
719
70-Gene Signature in Early Breast Cancer
clinical and genomic results were advised not to
receive adjuvant chemotherapy, whereas patients
who were categorized as having high-risk dis-
ease by both tests were advised to receive chemo-
therapy.
Patients with discordant results (i.e., either
high clinical risk and low genomic risk or low
clinical risk and high genomic risk) were ran-
domly assigned to the chemotherapy group or the
no-chemotherapy group on the basis of either
the clinical result or the genomic result. The
treatment randomization used a minimization
technique that was stratified according to in-
stitution, risk group, hormone-receptor status
(ER-positive or progesterone [PR]-positive vs.
ER-negative and PR-negative), nodal involvement
(yes or no), age (<50 years vs. ≥50 years), HER2
status (HER2-positive vs. HER2-negative vs. un-
known), axillary treatment (sentinel node only vs.
dissection), and type of surgery (mastectomy
vs. breast conservation).
Additional (optional) randomizations were im-
plemented in which patients who were assigned
to receive adjuvant chemotherapy (either random-
ly because of discordant results or because of
high-risk concordance of both tests) could be
randomly assigned to receive an anthracycline-
containing regimen or a docetaxel-plus-capecita-
bine regimen. Similarly, patients with hormone-
receptor–positive breast cancer could undergo
further randomization to a tamoxifen–letrozole
regimen or a letrozole-only regimen. Details re-
garding the various therapy regimens are pro-
vided in the legend for Figure S7 in the Supple-
mentary Appendix.
Protocol Revisions
A change in the RNA-extraction solution that
was used in the calculation of the 70-gene sig-
nature (a change that was not communicated by
the manufacturer) caused a temporary shift in
the risk calculation from May 24, 2009, to Janu-
ary 30, 2010, at which time the issue was discov-
ered and rectified (Table S5 in the Supplemen-
tary Appendix). Because of this shift, 162 patients
who had been identified as being at high ge-
nomic risk were subsequently identified as being
at low genomic risk with the use of the correct
solution (Tables S2 and S4 in the Supplementary
Appendix). The retroactively recalibrated results
were communicated to the independent data and
safety monitoring committee, to all members
of the ethics committees, to the investigators
(who oversaw informing the patients), and to the
TRANSBIG ethics committee. The clinical effect
of this risk revision was that an additional 28
patients received chemotherapy before the re-
sults were corrected, although no patient was
undertreated (Table S4 in the Supplementary
Appendix). For 113 additional patients, the des-
ignations of clinical or genomic risk were cor-
rected after enrollment, owing mainly to incor-
rect reporting of tumor characteristics at the
time of enrollment (Tables S2 and S3 in the
Supplementary Appendix). The actual risk after
the correction of all types of errors is referred to
as the “corrected risk.
The sample size was modified during the
trial from 6000 patients to 6600 patients because
the proportion of patients who were designated
as being at low clinical and genomic risk was
higher than was initially projected and because
of the need to compensate for the change in
solution used in RNA extraction.
Study End Points
The primary end point was survival without dis-
tant metastasis (event-free rate at 5 years), as
assessed in a time-to-event analysis. Secondary
end points were the proportion of patients who
received chemotherapy according to the clinical
risk as compared with the genomic risk as well
as overall survival and disease-free survival, as
assessed in time-to-event analyses. Survival with-
out distant metastasis was defined as the time
until the first distant metastatic recurrence or
death from any cause. Disease-free survival was
defined as the time until first disease progres-
sion (locoregional, distant relapse, ipsilateral or
contralateral invasive breast cancer, ductal carci-
noma in situ, or an invasive second primary
cancer) or death from any cause. Overall sur-
vival was defined as the time until death from
any cause. Data for patients who had no event at
the cutoff date for the final analysis were cen-
sored at the time of the last disease assessment
for survival without distant metastasis and for
disease-free survival and at the last follow-up
date for overall survival.
Study Oversight
The trial was overseen by the independent data
and safety monitoring committee of the EORTC.
A logistics pilot study validated the real-time
centralized analysis of frozen samples in several
European countries.
19
The MINDACT pilot phase
The New England Journal of Medicine
Downloaded from nejm.org at IMPERIAL COLLEGE LONDON on September 1, 2016. For personal use only. No other uses without permission.
Copyright © 2016 Massachusetts Medical Society. All rights reserved.

n engl j med 375;8 nejm.org August 25, 2016
720
The
new england journal
of
medicine
(involving the first 800 enrolled patients) con-
firmed the feasibility of the study.
21
Central
pathological review revealed high concordance
between local and central assessments
22,23
(Table
S12 in the Supplementary Appendix).
All randomizations were performed centrally,
initially at the International Drug Development
Institute and, as of 2010, at the EORTC. Genom-
ic profiling was performed by Agendia. The
drugs that were administered during the study
were provided by Novartis, Sanofi-Aventis, and
F. Hoffmann–La Roche, which had no other role
in the study, were not involved in the collection
or analysis of the data, and did not contribute to
the writing of the manuscript. Data collection
and statistical analyses were performed at the
EORTC. The analyses and the final version of the
manuscript were approved by all the authors,
who vouch for the accuracy and completeness of
the data and the adherence of the study to the
protocol.
Statistical Analysis
The primary analysis was designed to test
whether, among patients with high-risk clinical
features and a low-risk gene-expression profile
who did not receive chemotherapy, the lower
boundary of the 95% confidence interval for the
rate of 5-year survival without distant metastasis
would be 92% (i.e., the noninferiority boundary)
or higher, at a one-sided significance level of
0.025. The primary test was to be performed
when two conditions were met: the standard
error for the rate of survival without distant metas-
tasis at 5 years was 0.01 or less and the percent-
age of patients in the primary-test population
with 5-year follow-up was 33% or more. A two-
sided 95% confidence interval for the 5-year rate
of survival without distant metastasis of more
than 92% was considered to indicate statistical
significance. Under these conditions, this test
has 80% power to reject the null hypothesis if
the true 5-year rate of survival without distant
metastasis is 95%.
In addition, three secondary analyses were
planned. In the first analysis, we evaluated the
outcomes in patients in the discordant-risk
groups according to whether they were assigned
to the chemotherapy group or the no-chemo-
therapy group. In the second analysis, we evalu-
ated outcomes in all patients according to
whether the use of chemotherapy had been rec-
ommended by either clinical risk or genomic
risk alone. To have an unbiased estimate for this
analysis, data for patients in the discordant-risk
groups were doubly weighted, because they were
underrepresented by a factor of two in the re-
sulting sample. In the third analysis, we calcu-
lated the percentage of all enrolled patients who
would be assigned to chemotherapy on the basis
of either clinical risk or genomic risk.
In the first two secondary analyses, we report
the results according to the risk category and
treatment assignment at the time of enrollment
(intention-to-treat population). In the third sec-
ondary analysis and in all analyses according to
risk group (namely, those for outcome, patient
characteristics, eligibility, and adherence), we
report the results according to the corrected
risk group.
Exploratory analyses that were defined pro-
spectively in the protocol or in the statistical
analysis plan or that were deemed to be of high
clinical relevance are so described in the text
and in the Supplementary Appendix. All analy-
ses were performed with the use of SAS soft-
ware, version 9.4 (SAS Institute).
Results
Study Patients
The cutoff date for the current analysis was
March 1, 2016; the median follow-up was 5.0
years. Between 2007 and 2011, a total of 11,288
patients underwent screening and 6693 were
enrolled in the study (Fig. S7 in the Supplemen-
tary Appendix). Of the 4595 patients (40.7%)
who underwent screening but were not enrolled,
the main reasons were the unsuitability of tu-
mor material for testing, a decision by the pa-
tient or an investigator not to participate in the
study, or other ineligibility factor (Table S1 in
the Supplementary Appendix).
The patients were divided into four main
groups, according to their clinical and genomic
risk: low clinical risk and low genomic risk,
which included 2745 patients (41.0%); low clini-
cal risk and high genomic risk, which included
592 patients (8.8%); high clinical risk and low
genomic risk, which included 1550 patients
(23.2%); and high clinical risk and high genomic
risk, which included 1806 patients (27.0%).
These numbers were calculated on the basis of
the corrected risk (Fig. 1, and Table S2 in the
The New England Journal of Medicine
Downloaded from nejm.org at IMPERIAL COLLEGE LONDON on September 1, 2016. For personal use only. No other uses without permission.
Copyright © 2016 Massachusetts Medical Society. All rights reserved.

n engl j med 375;8 nejm.org August 25, 2016
721
70-Gene Signature in Early Breast Cancer
Figure 1. Enrollment and Risk Groups Included in the Analyses.
Patients were categorized into four risk groups on the basis of the 70-gene signature to determine genomic risk and Adjuvant! Online to determine clinical risk. Patients were fur-
ther categorized according to their “corrected risk” after the discovery of risk changes due to factors including a change in the RNA-extraction solution while the study was ongoing
(genomic risk) and incorrect reporting of clinical risk at enrollment (clinical risk). In the analyses according to risk group (namely, those for outcome, patient characteristics, eligibil-
ity, and adherence), the results are reported according to the corrected risk. The primary analysis was conducted in the primary-test population. This population included patients
at high clinical risk and low genomic risk, who were randomly assigned to use the genomic risk for the decision to forgo chemotherapy and who adhered to the treatment assign-
ment of no chemotherapy. Patients with changes in clinical or genomic risk were excluded from the primary-test population. Treatment-randomization analyses for the groups with
discordant clinical and genomic risks were performed with the use of the risk at enrollment (intention-to-treat population). In the intention-to-treat population, patients were ana-
lyzed according to the randomized group, irrespective of adherence. The treatment-randomization analyses were repeated in the per-protocol population, which excluded patients
who were ineligible, had a change in their clinical or genomic risk, or were nonadherent to the treatment assignment. (Patients could have more than one reason for exclusion.)
Per-Protocol Population
Primary-
test
Population
6693 Women with invasive early-stage breast
cancer were enrolled
1873 Had high clinical risk and high
genomic risk at enrollment
1806 Had high clinical risk and high
genomic risk after correction
2634 Had low clinical risk and low
genomic risk at enrollment
2745 Had low clinical risk and low
genomic risk after correction
690 Had low clinical risk and high
genomic risk at enrollment
592 Had low clinical risk and high
genomic risk after correction
1497 Had high clinical risk and low
genomic risk at enrollment
1550 Had high clinical risk and low
genomic risk after correction
690 Underwent randomization 1497 Underwent randomization
4 Were ineligible
53 Had a change
in risk
42 Received chemo-
therapy
4 Were ineligible
57 Had a change
in risk
76 Did not receive
chemotherapy
5 Had unknown
chemotherapy
status
344 Were assigned to
receive chemotherapy
346 Were not assigned
to receive chemotherapy
254 Were included in
the per-protocol
population
224 Were included in
the per-protocol
population
12 Were ineligible
21 Had a change
in risk
85 Received chemo-
therapy
1 Had unknown
chemotherapy
status
11 Were ineligible
26 Had a change
in risk
128 Did not receive
chemotherapy
9 Had unknown
chemotherapy
status
749 Were assigned to
receive chemotherapy
748 Were not assigned
to receive chemotherapy
636 Were included in
the per-protocol
population
21 Had a change
in risk
85 Received chemo-
therapy
1 Had unknown
chemotherapy
status
644 Were included in
the primary-test
population
592 Were included in
the per-protocol
population
Intention-to-Treat Population
The New England Journal of Medicine
Downloaded from nejm.org at IMPERIAL COLLEGE LONDON on September 1, 2016. For personal use only. No other uses without permission.
Copyright © 2016 Massachusetts Medical Society. All rights reserved.

Citations
More filters
Journal ArticleDOI
22 Jan 2019-JAMA
TL;DR: This review focuses on current approaches and evolving strategies for local and systemic therapy of breast cancer as well as distinct risk profiles and treatment strategies.
Abstract: Importance Breast cancer will be diagnosed in 12% of women in the United States over the course of their lifetimes and more than 250 000 new cases of breast cancer were diagnosed in the United States in 2017. This review focuses on current approaches and evolving strategies for local and systemic therapy of breast cancer. Observations Breast cancer is categorized into 3 major subtypes based on the presence or absence of molecular markers for estrogen or progesterone receptors and human epidermal growth factor 2 (ERBB2; formerlyHER2): hormone receptor positive/ERBB2 negative (70% of patients),ERBB2positive (15%-20%), and triple-negative (tumors lacking all 3 standard molecular markers; 15%). More than 90% of breast cancers are not metastatic at the time of diagnosis. For people presenting without metastatic disease, therapeutic goals are tumor eradication and preventing recurrence. Triple-negative breast cancer is more likely to recur than the other 2 subtypes, with 85% 5-year breast cancer–specific survival for stage I triple-negative tumors vs 94% to 99% for hormone receptor positive andERBB2positive. Systemic therapy for nonmetastatic breast cancer is determined by subtype: patients with hormone receptor–positive tumors receive endocrine therapy, and a minority receive chemotherapy as well; patients withERBB2-positive tumors receiveERBB2-targeted antibody or small-molecule inhibitor therapy combined with chemotherapy; and patients with triple-negative tumors receive chemotherapy alone. Local therapy for all patients with nonmetastatic breast cancer consists of surgical resection, with consideration of postoperative radiation if lumpectomy is performed. Increasingly, some systemic therapy is delivered before surgery. Tailoring postoperative treatment based on preoperative treatment response is under investigation. Metastatic breast cancer is treated according to subtype, with goals of prolonging life and palliating symptoms. Median overall survival for metastatic triple-negative breast cancer is approximately 1 year vs approximately 5 years for the other 2 subtypes. Conclusions and Relevance Breast cancer consists of 3 major tumor subtypes categorized according to estrogen or progesterone receptor expression andERBB2gene amplification. The 3 subtypes have distinct risk profiles and treatment strategies. Optimal therapy for each patient depends on tumor subtype, anatomic cancer stage, and patient preferences.

2,310 citations

Journal ArticleDOI
TL;DR: This work presents the results of a meta-analysis conducted at the 2016 European Oncology and Radiotherapy Guidelines Working Group (ESMO) workshop on breast cancer diagnosis and prognosis of women with atypical central giant cell granuloma (CGM) who have previously had surgery.

2,274 citations

Journal ArticleDOI
TL;DR: In this paper, the authors performed a prospective trial involving 10,273 women with hormone-receptor-positive, human epidermal growth factor receptor 2 (HER2)-negative, axillary node-negative breast cancer.
Abstract: Background The recurrence score based on the 21-gene breast cancer assay predicts chemotherapy benefit if it is high and a low risk of recurrence in the absence of chemotherapy if it is low; however, there is uncertainty about the benefit of chemotherapy for most patients, who have a midrange score. Methods We performed a prospective trial involving 10,273 women with hormone-receptor–positive, human epidermal growth factor receptor 2 (HER2)–negative, axillary node–negative breast cancer. Of the 9719 eligible patients with follow-up information, 6711 (69%) had a midrange recurrence score of 11 to 25 and were randomly assigned to receive either chemoendocrine therapy or endocrine therapy alone. The trial was designed to show noninferiority of endocrine therapy alone for invasive disease–free survival (defined as freedom from invasive disease recurrence, second primary cancer, or death). Results Endocrine therapy was noninferior to chemoendocrine therapy in the analysis of invasive disease–free surv...

1,337 citations

Journal ArticleDOI
TL;DR: How different forms of liquid biopsies can be exploited to guide patient care and should ultimately be integrated into clinical practice is examined, focusing on liquid biopsy of ctDNA — arguably the most clinically advanced approach.
Abstract: During cancer progression and treatment, multiple subclonal populations of tumour cells compete with one another, with selective pressures leading to the emergence of predominant subclones that replicate and spread most proficiently, and are least susceptible to treatment. At present, the molecular landscapes of solid tumours are established using surgical or biopsy tissue samples. Tissue-based tumour profiles are, however, subject to sampling bias, provide only a snapshot of tumour heterogeneity, and cannot be obtained repeatedly. Genomic profiles of circulating cell-free tumour DNA (ctDNA) have been shown to closely match those of the corresponding tumours, with important implications for both molecular pathology and clinical oncology. Analyses of circulating nucleic acids, commonly referred to as 'liquid biopsies', can be used to monitor response to treatment, assess the emergence of drug resistance, and quantify minimal residual disease. In addition to blood, several other body fluids, such as urine, saliva, pleural effusions, and cerebrospinal fluid, can contain tumour-derived genetic information. The molecular profiles gathered from ctDNA can be further complemented with those obtained through analysis of circulating tumour cells (CTCs), as well as RNA, proteins, and lipids contained within vesicles, such as exosomes. In this Review, we examine how different forms of liquid biopsies can be exploited to guide patient care and should ultimately be integrated into clinical practice, focusing on liquid biopsy of ctDNA - arguably the most clinically advanced approach.

1,292 citations

Journal ArticleDOI
TL;DR: The 15th St. Gallen International Breast Cancer Conference 2017 in Vienna, Austria reviewed substantial new evidence on loco-regional and systemic therapies for early breast cancer, and recommended bisphosphonate use in postmenopausal women to prevent breast cancer recurrence.

777 citations

References
More filters
Journal ArticleDOI
TL;DR: Survival analyses on a subcohort of patients with locally advanced breast cancer uniformly treated in a prospective study showed significantly different outcomes for the patients belonging to the various groups, including a poor prognosis for the basal-like subtype and a significant difference in outcome for the two estrogen receptor-positive groups.
Abstract: The purpose of this study was to classify breast carcinomas based on variations in gene expression patterns derived from cDNA microarrays and to correlate tumor characteristics to clinical outcome. A total of 85 cDNA microarray experiments representing 78 cancers, three fibroadenomas, and four normal breast tissues were analyzed by hierarchical clustering. As reported previously, the cancers could be classified into a basal epithelial-like group, an ERBB2-overexpressing group and a normal breast-like group based on variations in gene expression. A novel finding was that the previously characterized luminal epithelial/estrogen receptor-positive group could be divided into at least two subgroups, each with a distinctive expression profile. These subtypes proved to be reasonably robust by clustering using two different gene sets: first, a set of 456 cDNA clones previously selected to reflect intrinsic properties of the tumors and, second, a gene set that highly correlated with patient outcome. Survival analyses on a subcohort of patients with locally advanced breast cancer uniformly treated in a prospective study showed significantly different outcomes for the patients belonging to the various groups, including a poor prognosis for the basal-like subtype and a significant difference in outcome for the two estrogen receptor-positive groups.

10,791 citations

Journal ArticleDOI
31 Jan 2002-Nature
TL;DR: DNA microarray analysis on primary breast tumours of 117 young patients is used and supervised classification is applied to identify a gene expression signature strongly predictive of a short interval to distant metastases (‘poor prognosis’ signature) in patients without tumour cells in local lymph nodes at diagnosis, providing a strategy to select patients who would benefit from adjuvant therapy.
Abstract: Breast cancer patients with the same stage of disease can have markedly different treatment responses and overall outcome. The strongest predictors for metastases (for example, lymph node status and histological grade) fail to classify accurately breast tumours according to their clinical behaviour. Chemotherapy or hormonal therapy reduces the risk of distant metastases by approximately one-third; however, 70-80% of patients receiving this treatment would have survived without it. None of the signatures of breast cancer gene expression reported to date allow for patient-tailored therapy strategies. Here we used DNA microarray analysis on primary breast tumours of 117 young patients, and applied supervised classification to identify a gene expression signature strongly predictive of a short interval to distant metastases ('poor prognosis' signature) in patients without tumour cells in local lymph nodes at diagnosis (lymph node negative). In addition, we established a signature that identifies tumours of BRCA1 carriers. The poor prognosis signature consists of genes regulating cell cycle, invasion, metastasis and angiogenesis. This gene expression profile will outperform all currently used clinical parameters in predicting disease outcome. Our findings provide a strategy to select patients who would benefit from adjuvant therapy.

9,664 citations

Journal ArticleDOI
TL;DR: The gene-expression profile studied is a more powerful predictor of the outcome of disease in young patients with breast cancer than standard systems based on clinical and histologic criteria.
Abstract: Background A more accurate means of prognostication in breast cancer will improve the selection of patients for adjuvant systemic therapy. Methods Using microarray analysis to evaluate our previously established 70-gene prognosis profile, we classified a series of 295 consecutive patients with primary breast carcinomas as having a gene-expression signature associated with either a poor prognosis or a good prognosis. All patients had stage I or II breast cancer and were younger than 53 years old; 151 had lymph-node–negative disease, and 144 had lymph-node–positive disease. We evaluated the predictive power of the prognosis profile using univariable and multivariable statistical analyses. Results Among the 295 patients, 180 had a poor-prognosis signature and 115 had a good-prognosis signature, and the mean (±SE) overall 10-year survival rates were 54.6±4.4 percent and 94.5±2.6 percent, respectively. At 10 years, the probability of remaining free of distant metastases was 50.6±4.5 percent in the group with a...

5,902 citations

Journal ArticleDOI
TL;DR: The recurrence score has been validated as quantifying the likelihood of distant recurrence in tamoxifen-treated patients with node-negative, estrogen-receptor-positive breast cancer and could be used as a continuous function to predict distant recurrent in individual patients.
Abstract: background The likelihood of distant recurrence in patients with breast cancer who have no involved lymph nodes and estrogen-receptor–positive tumors is poorly defined by clinical and histopathological measures. methods We tested whether the results of a reverse-transcriptase–polymerase-chain-reaction (RT-PCR) assay of 21 prospectively selected genes in paraffin-embedded tumor tissue would correlate with the likelihood of distant recurrence in patients with node-negative, tamoxifen-treated breast cancer who were enrolled in the National Surgical Adjuvant Breast and Bowel Project clinical trial B-14. The levels of expression of 16 cancerrelated genes and 5 reference genes were used in a prospectively defined algorithm to calculate a recurrence score and to determine a risk group (low, intermediate, or high) for each patient. results Adequate RT-PCR profiles were obtained in 668 of 675 tumor blocks. The proportions of patients categorized as having a low, intermediate, or high risk by the RT-PCR assay were 51, 22, and 27 percent, respectively. The Kaplan–Meier estimates of the rates of distant recurrence at 10 years in the low-risk, intermediate-risk, and high-risk groups were 6.8 percent (95 percent confidence interval, 4.0 to 9.6), 14.3 percent (95 percent confidence interval, 8.3 to 20.3), and 30.5 percent (95 percent confidence interval, 23.6 to 37.4). The rate in the low-risk group was significantly lower than that in the high-risk group (P<0.001). In a multivariate Cox model, the recurrence score provided significant predictive power that was independent of age and tumor size (P<0.001). The recurrence score was also predictive of overall survival (P<0.001) and could be used as a continuous function to predict distant recurrence in individual patients. conclusions The recurrence score has been validated as quantifying the likelihood of distant recurrence in tamoxifen-treated patients with node-negative, estrogen-receptor–positive breast cancer.

5,685 citations

Journal ArticleDOI
TL;DR: In meta-analyses involving taxane-based or anthracycline-based regimens, proportional risk reductions were little affected by age, nodal status, tumour diameter or differentiation (moderate or poor; few were well differentiated), oestrogen receptor status, or tamoxifen use.

1,647 citations

Related Papers (5)
Frequently Asked Questions (14)
Q1. What are the contributions in "70-gene signature as an aid to treatment decisions in early-stage breast cancer" ?

In this randomized, phase 3 study, the authors enrolled 6693 women with early-stage breast cancer and determined their genomic risk ( using the 70-gene signature ) and their clinical risk ( using a modified version of Adjuvant ! 

The primary goal was to assess whether, among patients with high-risk clinical features and a low-risk gene-expression profile who did not receive chemotherapy, the lower boundary of the 95% confidence interval for the rate of 5-year survival without distant metastasis would be 92% (i.e., the noninferiority boundary) or higher. 

At 5 years, the rate of survival without distant metastasis in this group was 94.7% (95% confidence interval, 92.5 to 96.2) among those not receiving chemotherapy. 

An independent validation study by the TRANSBIG consortium, a network of some 40 partners in 21 countries associated with the Breast International Group (BIG), confirmed that the 70-gene signature, which has been approved by the Food and Drug Administration (FDA), is able to distinguish patients who are at significant risk for distant relapse and death from those at low risk. 

All rights reserved.n engl j med 375;8 nejm.org August 25, 2016718Women with early-stage breast cancer are often treated with adjuvant systemic therapy consisting of chemotherapy, endocrine therapy, agents against human epidermal growth factor receptor 2 (HER2), or combinations of these drugs when appropriate. 

In this international, prospective, randomized, phase 3 study, called the Microarray in NodeNegative and 1 to 3 Positive Lymph Node Disease May Avoid Chemotherapy (EORTC 10041/ BIG 3-04 MINDACT) study, the authors sought to provide evidence of the clinical utility13-15 of the addition of the 70-gene signature to standard clinical– pathological criteria in selecting patients for adjuvant chemotherapy. 

Given these findings, approximately 46% of women with breast cancer who are at high clinical risk might not require chemotherapy. 

The authors sought to provide prospective evidence of the clinical utility of the addition of the 70-gene signature to standard clinical–pathological criteria in selecting patients for adjuvant chemotherapy. 

Treatment decisions are based on characteristics of the tumor (hormonal receptor and HER2 status, tumor grade and size, and lymph-node status) and of the patient (age, menopausal status, and performance status). 

In 2007, respondents to an international survey identified the use of molecular signatures to select patients who could be spared adjuvant therapy as a high priority. 

Several genomic tests have been developed to better predict clinical outcome and to determine whether the addition of adjuvant chemotherapy to endocrine therapy is worthwhile. 

One such test, the 70-gene signature10 (MammaPrint), classifies tumors into groups that are associated with a good prognosis or a poor prognosis on the basis of the risk of distant recurrence at 5 years and at 10 years. 

Among women with early-stage breast cancer who were at high clinical risk and low genomic risk for recurrence, the receipt of no chemotherapy on the basis of the 70-gene signature led to a 5-year rate of survival without distant metastasis that was 1.5 percentage points lower than the rate with chemotherapy. 

Women at low clinical and genomic risk did not receive chemotherapy, whereas those at high clinical and genomic risk did receive such therapy.